User login
Longer interval between surgery and ET worsens survival outcomes in HR+ BC
Key clinical point: Delay in adjuvant endocrine therapy (ET) worsened survival outcomes in patients with hormone receptor-positive (HR+) breast cancer (BC) who received adjuvant radiotherapy after neoadjuvant chemotherapy.
Major finding: An interval of >14 weeks between surgery and receipt of ET was associated with worse recurrence-free survival (hazard ratio 3.20; P = .02).
Study details: This study analyzed 179 patients with HR+ BC from a multi-institutional database who received adjuvant radiotherapy, of which 68 patients received adjuvant ET before or during radiotherapy and 111 patients received ET after cessation of radiotherapy.
Disclosures: This study did not receive any funding. The authors declared no conflicts of interest.
Source: Sutton TL et al. Delayed adjuvant endocrine therapy is associated with decreased recurrence-free survival following neoadjuvant chemotherapy for breast cancer. Am J Surg. 2023 (Feb 24). Doi: 10.1016/j.amjsurg.2023.02.020
Key clinical point: Delay in adjuvant endocrine therapy (ET) worsened survival outcomes in patients with hormone receptor-positive (HR+) breast cancer (BC) who received adjuvant radiotherapy after neoadjuvant chemotherapy.
Major finding: An interval of >14 weeks between surgery and receipt of ET was associated with worse recurrence-free survival (hazard ratio 3.20; P = .02).
Study details: This study analyzed 179 patients with HR+ BC from a multi-institutional database who received adjuvant radiotherapy, of which 68 patients received adjuvant ET before or during radiotherapy and 111 patients received ET after cessation of radiotherapy.
Disclosures: This study did not receive any funding. The authors declared no conflicts of interest.
Source: Sutton TL et al. Delayed adjuvant endocrine therapy is associated with decreased recurrence-free survival following neoadjuvant chemotherapy for breast cancer. Am J Surg. 2023 (Feb 24). Doi: 10.1016/j.amjsurg.2023.02.020
Key clinical point: Delay in adjuvant endocrine therapy (ET) worsened survival outcomes in patients with hormone receptor-positive (HR+) breast cancer (BC) who received adjuvant radiotherapy after neoadjuvant chemotherapy.
Major finding: An interval of >14 weeks between surgery and receipt of ET was associated with worse recurrence-free survival (hazard ratio 3.20; P = .02).
Study details: This study analyzed 179 patients with HR+ BC from a multi-institutional database who received adjuvant radiotherapy, of which 68 patients received adjuvant ET before or during radiotherapy and 111 patients received ET after cessation of radiotherapy.
Disclosures: This study did not receive any funding. The authors declared no conflicts of interest.
Source: Sutton TL et al. Delayed adjuvant endocrine therapy is associated with decreased recurrence-free survival following neoadjuvant chemotherapy for breast cancer. Am J Surg. 2023 (Feb 24). Doi: 10.1016/j.amjsurg.2023.02.020
No increase in mortality risk with estrogen therapy in HR+ BC
Key clinical point: Estrogen therapy with or without the concurrent use of adjuvant aromatase inhibitors (AI) or tamoxifen did not increase the risk for mortality in women with early hormone receptor-positive (HR+) breast cancer (BC).
Major finding: No association was observed between BC mortality risk and receipt of estrogen therapy concurrent with AI, tamoxifen, or both AI and tamoxifen, although estrogen therapy without concurrent AI or tamoxifen was associated with decreased BC mortality risk (adjusted odds ratio 0.61; 95% CI 0.43-0.87).
Study details: Findings are from a population-based nested case-control study including patients with HR+ BC who received local estrogen therapy or AI, tamoxifen, or AI and tamoxifen sequentially, of which 1262 women died due to BC and were matched to 12,620 alive control individuals.
Disclosures: This study was supported by Bröstcancerförbundet, Sweden,and ALF Funding Region Örebro County, Sweden. The authors declared no conflicts of interest.
Source: Sund M et al. Estrogen therapy after breast cancer diagnosis and breast cancer mortality risk. Breast Cancer Res Treat. 2023 (Feb 11). Doi: 10.1007/s10549-023-06871-w
Key clinical point: Estrogen therapy with or without the concurrent use of adjuvant aromatase inhibitors (AI) or tamoxifen did not increase the risk for mortality in women with early hormone receptor-positive (HR+) breast cancer (BC).
Major finding: No association was observed between BC mortality risk and receipt of estrogen therapy concurrent with AI, tamoxifen, or both AI and tamoxifen, although estrogen therapy without concurrent AI or tamoxifen was associated with decreased BC mortality risk (adjusted odds ratio 0.61; 95% CI 0.43-0.87).
Study details: Findings are from a population-based nested case-control study including patients with HR+ BC who received local estrogen therapy or AI, tamoxifen, or AI and tamoxifen sequentially, of which 1262 women died due to BC and were matched to 12,620 alive control individuals.
Disclosures: This study was supported by Bröstcancerförbundet, Sweden,and ALF Funding Region Örebro County, Sweden. The authors declared no conflicts of interest.
Source: Sund M et al. Estrogen therapy after breast cancer diagnosis and breast cancer mortality risk. Breast Cancer Res Treat. 2023 (Feb 11). Doi: 10.1007/s10549-023-06871-w
Key clinical point: Estrogen therapy with or without the concurrent use of adjuvant aromatase inhibitors (AI) or tamoxifen did not increase the risk for mortality in women with early hormone receptor-positive (HR+) breast cancer (BC).
Major finding: No association was observed between BC mortality risk and receipt of estrogen therapy concurrent with AI, tamoxifen, or both AI and tamoxifen, although estrogen therapy without concurrent AI or tamoxifen was associated with decreased BC mortality risk (adjusted odds ratio 0.61; 95% CI 0.43-0.87).
Study details: Findings are from a population-based nested case-control study including patients with HR+ BC who received local estrogen therapy or AI, tamoxifen, or AI and tamoxifen sequentially, of which 1262 women died due to BC and were matched to 12,620 alive control individuals.
Disclosures: This study was supported by Bröstcancerförbundet, Sweden,and ALF Funding Region Örebro County, Sweden. The authors declared no conflicts of interest.
Source: Sund M et al. Estrogen therapy after breast cancer diagnosis and breast cancer mortality risk. Breast Cancer Res Treat. 2023 (Feb 11). Doi: 10.1007/s10549-023-06871-w
Preoperative breast MRI has no impact on survival in breast cancer
Key clinical point: Preoperative magnetic resonance imaging (MRI) of the breast increased mastectomy rates but did not affect the local recurrence and overall survival (OS) rates in patients with breast cancer (BC) who were eligible for breast-conserving surgery (BCS).
Major finding: Among BCS-eligible patients, surgical procedure was changed to mastectomy in 8.3% vs 0.4% of patients in the MRI vs routine radiologic exam group, respectively. There was no difference in local recurrence-free survival (hazard ratio [HR] 0.72; log-rank test P = .7) and OS (HR 1.37; log-rank test P = .8) between both patient populations.
Study details: Findings are from the phase 3 BREAST-MRI study including 524 patients with stage 0-III BC who were eligible for BCS and were randomly assigned to undergo preoperative MRI or radiologic exam routine with mammography and ultrasound.
Disclosures: This publication was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo, Brazil (FAPESP 2018/24224-9). The authors declared no conflicts of interest.
Source: Mota BS et al. Effects of preoperative magnetic resonance image on survival rates and surgical planning in breast cancer conservative surgery: Randomized controlled trial (BREAST-MRI trial). Breast Cancer Res Treat. 2023 (Feb 14). Doi: 10.1007/s10549-023-06884-5
Key clinical point: Preoperative magnetic resonance imaging (MRI) of the breast increased mastectomy rates but did not affect the local recurrence and overall survival (OS) rates in patients with breast cancer (BC) who were eligible for breast-conserving surgery (BCS).
Major finding: Among BCS-eligible patients, surgical procedure was changed to mastectomy in 8.3% vs 0.4% of patients in the MRI vs routine radiologic exam group, respectively. There was no difference in local recurrence-free survival (hazard ratio [HR] 0.72; log-rank test P = .7) and OS (HR 1.37; log-rank test P = .8) between both patient populations.
Study details: Findings are from the phase 3 BREAST-MRI study including 524 patients with stage 0-III BC who were eligible for BCS and were randomly assigned to undergo preoperative MRI or radiologic exam routine with mammography and ultrasound.
Disclosures: This publication was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo, Brazil (FAPESP 2018/24224-9). The authors declared no conflicts of interest.
Source: Mota BS et al. Effects of preoperative magnetic resonance image on survival rates and surgical planning in breast cancer conservative surgery: Randomized controlled trial (BREAST-MRI trial). Breast Cancer Res Treat. 2023 (Feb 14). Doi: 10.1007/s10549-023-06884-5
Key clinical point: Preoperative magnetic resonance imaging (MRI) of the breast increased mastectomy rates but did not affect the local recurrence and overall survival (OS) rates in patients with breast cancer (BC) who were eligible for breast-conserving surgery (BCS).
Major finding: Among BCS-eligible patients, surgical procedure was changed to mastectomy in 8.3% vs 0.4% of patients in the MRI vs routine radiologic exam group, respectively. There was no difference in local recurrence-free survival (hazard ratio [HR] 0.72; log-rank test P = .7) and OS (HR 1.37; log-rank test P = .8) between both patient populations.
Study details: Findings are from the phase 3 BREAST-MRI study including 524 patients with stage 0-III BC who were eligible for BCS and were randomly assigned to undergo preoperative MRI or radiologic exam routine with mammography and ultrasound.
Disclosures: This publication was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo, Brazil (FAPESP 2018/24224-9). The authors declared no conflicts of interest.
Source: Mota BS et al. Effects of preoperative magnetic resonance image on survival rates and surgical planning in breast cancer conservative surgery: Randomized controlled trial (BREAST-MRI trial). Breast Cancer Res Treat. 2023 (Feb 14). Doi: 10.1007/s10549-023-06884-5
Vaginal laser treatment not superior to sham laser therapy for genitourinary syndrome of menopause in BC survivors
Key clinical point: Vaginal laser treatment was not superior in improving sexual function and was less well tolerated than sham laser therapy (SLT) in survivors of breast cancer (BC) with genitourinary syndrome of menopause (GSM) who received treatment with aromatase inhibitors (AI).
Major finding: At 6 months, patients receiving fractional carbon dioxide laser therapy (CLT) and those receiving SLT had similar improvement in the female sexual function index score (P = .15), with the CLT vs SLT group having a significantly lower mean tolerance score (3.3 vs 4.1; P = .007).
Study details: Findings are from the randomized clinical trial LIGHT including 84 patients with BC who were receiving AI for GSM and were randomly assigned to receive a first-line therapy based on nonhormonal moisturizers and vaginal vibrator stimulation with fractional CLT or SLT.
Disclosures: This study was funded by grants from Instituto de Salud Carlos III and the European Union, with various items provided by DEKA, IntherPharma, CumLaude Lab, and BCNatal. The authors declared no conflicts of interest.
Source: Mension E et al. Effect of fractional carbon dioxide vs sham laser on sexual function in survivors of breast cancer receiving aromatase inhibitors for genitourinary syndrome of menopause: The LIGHT randomized clinical trial. JAMA Netw Open. 2023;6(2):e2255697 (Feb 10). Doi: 10.1001/jamanetworkopen.2022.55697
Key clinical point: Vaginal laser treatment was not superior in improving sexual function and was less well tolerated than sham laser therapy (SLT) in survivors of breast cancer (BC) with genitourinary syndrome of menopause (GSM) who received treatment with aromatase inhibitors (AI).
Major finding: At 6 months, patients receiving fractional carbon dioxide laser therapy (CLT) and those receiving SLT had similar improvement in the female sexual function index score (P = .15), with the CLT vs SLT group having a significantly lower mean tolerance score (3.3 vs 4.1; P = .007).
Study details: Findings are from the randomized clinical trial LIGHT including 84 patients with BC who were receiving AI for GSM and were randomly assigned to receive a first-line therapy based on nonhormonal moisturizers and vaginal vibrator stimulation with fractional CLT or SLT.
Disclosures: This study was funded by grants from Instituto de Salud Carlos III and the European Union, with various items provided by DEKA, IntherPharma, CumLaude Lab, and BCNatal. The authors declared no conflicts of interest.
Source: Mension E et al. Effect of fractional carbon dioxide vs sham laser on sexual function in survivors of breast cancer receiving aromatase inhibitors for genitourinary syndrome of menopause: The LIGHT randomized clinical trial. JAMA Netw Open. 2023;6(2):e2255697 (Feb 10). Doi: 10.1001/jamanetworkopen.2022.55697
Key clinical point: Vaginal laser treatment was not superior in improving sexual function and was less well tolerated than sham laser therapy (SLT) in survivors of breast cancer (BC) with genitourinary syndrome of menopause (GSM) who received treatment with aromatase inhibitors (AI).
Major finding: At 6 months, patients receiving fractional carbon dioxide laser therapy (CLT) and those receiving SLT had similar improvement in the female sexual function index score (P = .15), with the CLT vs SLT group having a significantly lower mean tolerance score (3.3 vs 4.1; P = .007).
Study details: Findings are from the randomized clinical trial LIGHT including 84 patients with BC who were receiving AI for GSM and were randomly assigned to receive a first-line therapy based on nonhormonal moisturizers and vaginal vibrator stimulation with fractional CLT or SLT.
Disclosures: This study was funded by grants from Instituto de Salud Carlos III and the European Union, with various items provided by DEKA, IntherPharma, CumLaude Lab, and BCNatal. The authors declared no conflicts of interest.
Source: Mension E et al. Effect of fractional carbon dioxide vs sham laser on sexual function in survivors of breast cancer receiving aromatase inhibitors for genitourinary syndrome of menopause: The LIGHT randomized clinical trial. JAMA Netw Open. 2023;6(2):e2255697 (Feb 10). Doi: 10.1001/jamanetworkopen.2022.55697
No detrimental effect of ET on cognitive functioning in early BC
Key clinical point: Endocrine therapy (ET) did not have any detrimental effect on cognitive abilities in women aged ≥70 years with early breast cancer (BC).
Major finding: Patients receiving ET had a Mini-Mental State Examination (MMSE) score of 28.1, with mild and severe cognitive impairments observed in 25% and 2% of patients, respectively. The MSME score improved by 0.4 points (P = .013) after 15 months and by 0.5 points (P = .018) after 27 months in patients receiving ET.
Study details: Findings are from the observational CLIMB study including 273 women with stage I-III BC who were ≥70 years old, of which 48% received ET.
Disclosures: This study was funded by the KWF Dutch Cancer Society. The authors declared no conflicts of interest.
Source: Baltussen JC et al. Association between endocrine therapy and cognitive decline in older women with early breast cancer: Findings from the prospective CLIMB study. Eur J Cancer. 2023 (Feb 16). Doi: 10.1016/j.ejca.2023.02.008
Key clinical point: Endocrine therapy (ET) did not have any detrimental effect on cognitive abilities in women aged ≥70 years with early breast cancer (BC).
Major finding: Patients receiving ET had a Mini-Mental State Examination (MMSE) score of 28.1, with mild and severe cognitive impairments observed in 25% and 2% of patients, respectively. The MSME score improved by 0.4 points (P = .013) after 15 months and by 0.5 points (P = .018) after 27 months in patients receiving ET.
Study details: Findings are from the observational CLIMB study including 273 women with stage I-III BC who were ≥70 years old, of which 48% received ET.
Disclosures: This study was funded by the KWF Dutch Cancer Society. The authors declared no conflicts of interest.
Source: Baltussen JC et al. Association between endocrine therapy and cognitive decline in older women with early breast cancer: Findings from the prospective CLIMB study. Eur J Cancer. 2023 (Feb 16). Doi: 10.1016/j.ejca.2023.02.008
Key clinical point: Endocrine therapy (ET) did not have any detrimental effect on cognitive abilities in women aged ≥70 years with early breast cancer (BC).
Major finding: Patients receiving ET had a Mini-Mental State Examination (MMSE) score of 28.1, with mild and severe cognitive impairments observed in 25% and 2% of patients, respectively. The MSME score improved by 0.4 points (P = .013) after 15 months and by 0.5 points (P = .018) after 27 months in patients receiving ET.
Study details: Findings are from the observational CLIMB study including 273 women with stage I-III BC who were ≥70 years old, of which 48% received ET.
Disclosures: This study was funded by the KWF Dutch Cancer Society. The authors declared no conflicts of interest.
Source: Baltussen JC et al. Association between endocrine therapy and cognitive decline in older women with early breast cancer: Findings from the prospective CLIMB study. Eur J Cancer. 2023 (Feb 16). Doi: 10.1016/j.ejca.2023.02.008
Delaying BC surgery after diagnosis worsens survival
Key clinical point: Patients with breast cancer (BC) should be recommended to undergo primary breast surgery within 8 weeks from BC diagnosis for best survival outcomes.
Major finding: Overall survival outcomes were worsened by ≥15% when the interval between BC diagnosis and surgery was ≥9 weeks compared with 0-4 weeks (hazard ratio at 9 weeks 1.15; P < .001).
Study details: Findings are from a case series study including 373,334 female patients with stage I-III BC who underwent primary breast surgery.
Disclosures: This study was supported by grants from the US National Cancer Institute and other sources. LG Wilke declared being the founder of and stock owner in Elucent Medical. BM Hanlon declared receiving grants from the US National Institutes of Health outside the submitted work.
Source: Wiener AA et al. Reexamining time from breast cancer diagnosis to primary breast surgery. JAMA Surg. 2023 (Mar 1). Doi: 10.1001/jamasurg.2022.8388
Key clinical point: Patients with breast cancer (BC) should be recommended to undergo primary breast surgery within 8 weeks from BC diagnosis for best survival outcomes.
Major finding: Overall survival outcomes were worsened by ≥15% when the interval between BC diagnosis and surgery was ≥9 weeks compared with 0-4 weeks (hazard ratio at 9 weeks 1.15; P < .001).
Study details: Findings are from a case series study including 373,334 female patients with stage I-III BC who underwent primary breast surgery.
Disclosures: This study was supported by grants from the US National Cancer Institute and other sources. LG Wilke declared being the founder of and stock owner in Elucent Medical. BM Hanlon declared receiving grants from the US National Institutes of Health outside the submitted work.
Source: Wiener AA et al. Reexamining time from breast cancer diagnosis to primary breast surgery. JAMA Surg. 2023 (Mar 1). Doi: 10.1001/jamasurg.2022.8388
Key clinical point: Patients with breast cancer (BC) should be recommended to undergo primary breast surgery within 8 weeks from BC diagnosis for best survival outcomes.
Major finding: Overall survival outcomes were worsened by ≥15% when the interval between BC diagnosis and surgery was ≥9 weeks compared with 0-4 weeks (hazard ratio at 9 weeks 1.15; P < .001).
Study details: Findings are from a case series study including 373,334 female patients with stage I-III BC who underwent primary breast surgery.
Disclosures: This study was supported by grants from the US National Cancer Institute and other sources. LG Wilke declared being the founder of and stock owner in Elucent Medical. BM Hanlon declared receiving grants from the US National Institutes of Health outside the submitted work.
Source: Wiener AA et al. Reexamining time from breast cancer diagnosis to primary breast surgery. JAMA Surg. 2023 (Mar 1). Doi: 10.1001/jamasurg.2022.8388
Node-negative HER2+ BC: End-of-study analysis supports adjuvant treatment with paclitaxel and trastuzumab
Key clinical point: Adjuvant treatment with paclitaxel and trastuzumab demonstrated very good long-term survival outcomes in patients with small, node-negative, human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC).
Major finding: After a median follow-up of 10.8 years, only 31 invasive disease-free survival events occurred, of which six were locoregional ipsilateral recurrences, nine were new contralateral breast cancers, six were distant recurrences, and 10 were all-cause deaths. The 10-year invasive disease-free survival rate was >90% (91.3%; 95% CI 88.3%-94.4%).
Study details: Findings are from the phase 2 APT study including 406 patients with small (≤3 cm), node-negative, HER2+ BC who received adjuvant paclitaxel+trastuzumab for 12 weeks followed by trastuzumab for 40 weeks to complete a full year of trastuzumab treatment.
Disclosures: This study was funded by Genentech. The authors declared receiving consulting or advisory board fees, grant support, payment, or having other ties with several sources.
Source: Tolaney SM et al. Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer: Final 10-year analysis of the open-label, single-arm, phase 2 APT trial. Lancet Oncol. 2023;24(3):273-285 (Feb 27). Doi: 10.1016/S1470-2045(23)00051-7
Key clinical point: Adjuvant treatment with paclitaxel and trastuzumab demonstrated very good long-term survival outcomes in patients with small, node-negative, human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC).
Major finding: After a median follow-up of 10.8 years, only 31 invasive disease-free survival events occurred, of which six were locoregional ipsilateral recurrences, nine were new contralateral breast cancers, six were distant recurrences, and 10 were all-cause deaths. The 10-year invasive disease-free survival rate was >90% (91.3%; 95% CI 88.3%-94.4%).
Study details: Findings are from the phase 2 APT study including 406 patients with small (≤3 cm), node-negative, HER2+ BC who received adjuvant paclitaxel+trastuzumab for 12 weeks followed by trastuzumab for 40 weeks to complete a full year of trastuzumab treatment.
Disclosures: This study was funded by Genentech. The authors declared receiving consulting or advisory board fees, grant support, payment, or having other ties with several sources.
Source: Tolaney SM et al. Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer: Final 10-year analysis of the open-label, single-arm, phase 2 APT trial. Lancet Oncol. 2023;24(3):273-285 (Feb 27). Doi: 10.1016/S1470-2045(23)00051-7
Key clinical point: Adjuvant treatment with paclitaxel and trastuzumab demonstrated very good long-term survival outcomes in patients with small, node-negative, human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC).
Major finding: After a median follow-up of 10.8 years, only 31 invasive disease-free survival events occurred, of which six were locoregional ipsilateral recurrences, nine were new contralateral breast cancers, six were distant recurrences, and 10 were all-cause deaths. The 10-year invasive disease-free survival rate was >90% (91.3%; 95% CI 88.3%-94.4%).
Study details: Findings are from the phase 2 APT study including 406 patients with small (≤3 cm), node-negative, HER2+ BC who received adjuvant paclitaxel+trastuzumab for 12 weeks followed by trastuzumab for 40 weeks to complete a full year of trastuzumab treatment.
Disclosures: This study was funded by Genentech. The authors declared receiving consulting or advisory board fees, grant support, payment, or having other ties with several sources.
Source: Tolaney SM et al. Adjuvant paclitaxel and trastuzumab for node-negative, HER2-positive breast cancer: Final 10-year analysis of the open-label, single-arm, phase 2 APT trial. Lancet Oncol. 2023;24(3):273-285 (Feb 27). Doi: 10.1016/S1470-2045(23)00051-7
Alisertib shows promising antitumor activity with or without fulvestrant in endocrine-resistant metastatic BC
Key clinical point: Alisertib demonstrated clinically meaningful antitumor activity both with and without fulvestrant and a tolerable safety profile in patients with endocrine-resistant, human epidermal growth factor receptor 2-negative (ERBB2−) metastatic breast cancer (BC).
Major finding: The objective response rates were 19.6% (90% CI 10.6%-31.7%) and 20.0% (90% CI 10.9%-32.3%) for alisertib and alisertib+fulvestrant, respectively, and the median progression-free survival was approximately 5 months for both treatment arms. The most common grade ≥3 adverse events were neutropenia, leukopenia, and anemia.
Study details: Findings are from the phase 2 TBCRC041 trial including 91 patients with endocrine-resistant, ERBB2− metastatic BC who were previously treated with a cyclin-dependent kinase 4/6 inhibitor and were randomly assigned to receive alisertib alone or alisertib+fulvestrant.
Disclosures: This study was supported by Takeda Oncology. The authors declared receiving funding, fees, or research support or having other ties with several sources, including Takeda Oncology.
Source: Haddad TC et al. Evaluation of alisertib alone or combined with fulvestrant in patients with endocrine-resistant advanced breast cancer: The phase 2 TBCRC041 randomized clinical trial. JAMA Oncol. 2023 (Mar 9). Doi: 10.1001/jamaoncol.2022.7949
Key clinical point: Alisertib demonstrated clinically meaningful antitumor activity both with and without fulvestrant and a tolerable safety profile in patients with endocrine-resistant, human epidermal growth factor receptor 2-negative (ERBB2−) metastatic breast cancer (BC).
Major finding: The objective response rates were 19.6% (90% CI 10.6%-31.7%) and 20.0% (90% CI 10.9%-32.3%) for alisertib and alisertib+fulvestrant, respectively, and the median progression-free survival was approximately 5 months for both treatment arms. The most common grade ≥3 adverse events were neutropenia, leukopenia, and anemia.
Study details: Findings are from the phase 2 TBCRC041 trial including 91 patients with endocrine-resistant, ERBB2− metastatic BC who were previously treated with a cyclin-dependent kinase 4/6 inhibitor and were randomly assigned to receive alisertib alone or alisertib+fulvestrant.
Disclosures: This study was supported by Takeda Oncology. The authors declared receiving funding, fees, or research support or having other ties with several sources, including Takeda Oncology.
Source: Haddad TC et al. Evaluation of alisertib alone or combined with fulvestrant in patients with endocrine-resistant advanced breast cancer: The phase 2 TBCRC041 randomized clinical trial. JAMA Oncol. 2023 (Mar 9). Doi: 10.1001/jamaoncol.2022.7949
Key clinical point: Alisertib demonstrated clinically meaningful antitumor activity both with and without fulvestrant and a tolerable safety profile in patients with endocrine-resistant, human epidermal growth factor receptor 2-negative (ERBB2−) metastatic breast cancer (BC).
Major finding: The objective response rates were 19.6% (90% CI 10.6%-31.7%) and 20.0% (90% CI 10.9%-32.3%) for alisertib and alisertib+fulvestrant, respectively, and the median progression-free survival was approximately 5 months for both treatment arms. The most common grade ≥3 adverse events were neutropenia, leukopenia, and anemia.
Study details: Findings are from the phase 2 TBCRC041 trial including 91 patients with endocrine-resistant, ERBB2− metastatic BC who were previously treated with a cyclin-dependent kinase 4/6 inhibitor and were randomly assigned to receive alisertib alone or alisertib+fulvestrant.
Disclosures: This study was supported by Takeda Oncology. The authors declared receiving funding, fees, or research support or having other ties with several sources, including Takeda Oncology.
Source: Haddad TC et al. Evaluation of alisertib alone or combined with fulvestrant in patients with endocrine-resistant advanced breast cancer: The phase 2 TBCRC041 randomized clinical trial. JAMA Oncol. 2023 (Mar 9). Doi: 10.1001/jamaoncol.2022.7949
Antibody-Drug Conjugates: Targeted Treatments Providing Hope for Patients With Breast Cancer
The restrictive therapeutic index of chemotherapy has led to the emergence of antibody-drug conjugates (ADCs), medicines that combine the specificity of monoclonal antibodies (mAbs) with the cytotoxic effects of chemotherapy to deliver cytotoxic payloads to cancer cells. This targeted approach can reduce the side effects of chemotherapy and improve the effectiveness of treatment. Several ADCs, including ado-trastuzumab emtansine (Kadcyla), sacituzumab govitecan-hziy (Trodelvy), and fam-trastuzumab deruxtecan-nxki (Enhertu), are currently approved for treating some types of breast cancer (BC).
The ADC trastuzumab emtansine was approved specifically for treating human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer (mBC) in patients who have previously been treated with trastuzumab and a taxane (paclitaxel or docetaxel) and who have already been treated for mBC or have developed tumor recurrence within 6 months of receiving adjuvant therapy. The US Food and Drug Administration (FDA) approval was based on the results of the EMILIA study, a phase 3 clinical trial that compared treatment with trastuzumab emtansine vs capecitabine + lapatinib in participants with HER2+, locally advanced, or metastatic BC. This trial emerged from the need for well-tolerated, HER2-directed therapies for patients with this type of cancer. Trastuzumab emtansine consists of trastuzumab, a mAb that targets HER2 (which is overexpressed in about 20% of BCs), linked to emtansine, a cytotoxic payload that inhibits cell division. The trastuzumab emtansine group had a median overall survival (OS) of 29.9 months vs 25.9 months in the capecitabine + lapatinib group, for a hazard ratio (HR) of 0.75 (95% CI: 0.64, 0.88).
Another ADC, sacituzumab govitecan, targets the Trop-2 protein, which is overexpressed in BC. This ADC includes a mAb that is linked to SN-38, a cytotoxic payload that inhibits DNA replication. Triple-negative breast cancer (TNBC) is a subtype of BC that does not have receptors for estrogen, progesterone, or HER2—making it more difficult to treat than other forms of BC. Sacituzumab govitecan is used to treat patients with metastatic TNBC who have received at least 2 prior therapies for metastatic disease. Sacituzumab govitecan is also approved for the treatment of patients with unresectable locally advanced or metastatic hormone-receptor–positive (HR+), and HER2-negative (HER2−) BC who have received endocrine-based therapy and at least 2 additional systemic therapies in the metastatic setting. Sacituzumab govitecan was the first Trop-2–directed ADC to demonstrate OS benefit in patients with HR+/HER2− mBC who had received prior endocrine-based therapy and at least 2 chemotherapies. It is now also recommended as a Category 1 preferred treatment for metastatic HR+/HER2− BC by the National Comprehensive Cancer Network.
The results of the TROPiCS-02 study, which led to the FDA approval of sacituzumab govitecan, demonstrated a median OS of 14.4 months with sacituzumab govitecan vs 11.2 months with treatment of physician’s choice (HR, 0.79; 95% CI: 0.65, 0.96; P = 0.02). This represents a 3.2-month improvement in survival and a 21% reduction in the risk for patient death. Before this medicine was approved, there were limited options to offer patients with BC after endocrine-based therapy and chemotherapy.
A third ADC, trastuzumab deruxtecan, targets the HER2 protein, like trastuzumab emtansine, but with a different cytotoxic payload. It consists of trastuzumab linked to deruxtecan, whose cytotoxicity inhibits DNA replication. It is approved for the treatment of HER2+ mBC. Its FDA approval was based on the results of the DESTINY-Breast04 phase 3 clinical trial, which demonstrated that treatment with trastuzumab deruxtecan, when compared with standard-of-care chemotherapy, doubles progression-free survival among patients with mBC that expresses low levels of HER2. The median OS for patients in the HR+ group who received trastuzumab deruxtecan was 23.9 months vs 17.5 months for those who received chemotherapy. In the hormone receptor-negative (HR−) group, the median OS for those who took trastuzumab deruxtecan was 16.6 months vs 10.3 months for those treated with chemotherapy.
The emergence of ADCs have demonstrated promising advancements in the treatment of BC, particularly in patients with HER2+ or triple-negative disease. ADCs have given new hope to and prolonged life for patients living with pretreated HR+/HER2− mBC. ADCs also have the potential to provide a more effective and less toxic treatment option for patients with BC. However, further research is needed to fully understand their long-term effects and to develop new ADCs that target other types of BC.
The restrictive therapeutic index of chemotherapy has led to the emergence of antibody-drug conjugates (ADCs), medicines that combine the specificity of monoclonal antibodies (mAbs) with the cytotoxic effects of chemotherapy to deliver cytotoxic payloads to cancer cells. This targeted approach can reduce the side effects of chemotherapy and improve the effectiveness of treatment. Several ADCs, including ado-trastuzumab emtansine (Kadcyla), sacituzumab govitecan-hziy (Trodelvy), and fam-trastuzumab deruxtecan-nxki (Enhertu), are currently approved for treating some types of breast cancer (BC).
The ADC trastuzumab emtansine was approved specifically for treating human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer (mBC) in patients who have previously been treated with trastuzumab and a taxane (paclitaxel or docetaxel) and who have already been treated for mBC or have developed tumor recurrence within 6 months of receiving adjuvant therapy. The US Food and Drug Administration (FDA) approval was based on the results of the EMILIA study, a phase 3 clinical trial that compared treatment with trastuzumab emtansine vs capecitabine + lapatinib in participants with HER2+, locally advanced, or metastatic BC. This trial emerged from the need for well-tolerated, HER2-directed therapies for patients with this type of cancer. Trastuzumab emtansine consists of trastuzumab, a mAb that targets HER2 (which is overexpressed in about 20% of BCs), linked to emtansine, a cytotoxic payload that inhibits cell division. The trastuzumab emtansine group had a median overall survival (OS) of 29.9 months vs 25.9 months in the capecitabine + lapatinib group, for a hazard ratio (HR) of 0.75 (95% CI: 0.64, 0.88).
Another ADC, sacituzumab govitecan, targets the Trop-2 protein, which is overexpressed in BC. This ADC includes a mAb that is linked to SN-38, a cytotoxic payload that inhibits DNA replication. Triple-negative breast cancer (TNBC) is a subtype of BC that does not have receptors for estrogen, progesterone, or HER2—making it more difficult to treat than other forms of BC. Sacituzumab govitecan is used to treat patients with metastatic TNBC who have received at least 2 prior therapies for metastatic disease. Sacituzumab govitecan is also approved for the treatment of patients with unresectable locally advanced or metastatic hormone-receptor–positive (HR+), and HER2-negative (HER2−) BC who have received endocrine-based therapy and at least 2 additional systemic therapies in the metastatic setting. Sacituzumab govitecan was the first Trop-2–directed ADC to demonstrate OS benefit in patients with HR+/HER2− mBC who had received prior endocrine-based therapy and at least 2 chemotherapies. It is now also recommended as a Category 1 preferred treatment for metastatic HR+/HER2− BC by the National Comprehensive Cancer Network.
The results of the TROPiCS-02 study, which led to the FDA approval of sacituzumab govitecan, demonstrated a median OS of 14.4 months with sacituzumab govitecan vs 11.2 months with treatment of physician’s choice (HR, 0.79; 95% CI: 0.65, 0.96; P = 0.02). This represents a 3.2-month improvement in survival and a 21% reduction in the risk for patient death. Before this medicine was approved, there were limited options to offer patients with BC after endocrine-based therapy and chemotherapy.
A third ADC, trastuzumab deruxtecan, targets the HER2 protein, like trastuzumab emtansine, but with a different cytotoxic payload. It consists of trastuzumab linked to deruxtecan, whose cytotoxicity inhibits DNA replication. It is approved for the treatment of HER2+ mBC. Its FDA approval was based on the results of the DESTINY-Breast04 phase 3 clinical trial, which demonstrated that treatment with trastuzumab deruxtecan, when compared with standard-of-care chemotherapy, doubles progression-free survival among patients with mBC that expresses low levels of HER2. The median OS for patients in the HR+ group who received trastuzumab deruxtecan was 23.9 months vs 17.5 months for those who received chemotherapy. In the hormone receptor-negative (HR−) group, the median OS for those who took trastuzumab deruxtecan was 16.6 months vs 10.3 months for those treated with chemotherapy.
The emergence of ADCs have demonstrated promising advancements in the treatment of BC, particularly in patients with HER2+ or triple-negative disease. ADCs have given new hope to and prolonged life for patients living with pretreated HR+/HER2− mBC. ADCs also have the potential to provide a more effective and less toxic treatment option for patients with BC. However, further research is needed to fully understand their long-term effects and to develop new ADCs that target other types of BC.
The restrictive therapeutic index of chemotherapy has led to the emergence of antibody-drug conjugates (ADCs), medicines that combine the specificity of monoclonal antibodies (mAbs) with the cytotoxic effects of chemotherapy to deliver cytotoxic payloads to cancer cells. This targeted approach can reduce the side effects of chemotherapy and improve the effectiveness of treatment. Several ADCs, including ado-trastuzumab emtansine (Kadcyla), sacituzumab govitecan-hziy (Trodelvy), and fam-trastuzumab deruxtecan-nxki (Enhertu), are currently approved for treating some types of breast cancer (BC).
The ADC trastuzumab emtansine was approved specifically for treating human epidermal growth factor receptor 2 positive (HER2+) metastatic breast cancer (mBC) in patients who have previously been treated with trastuzumab and a taxane (paclitaxel or docetaxel) and who have already been treated for mBC or have developed tumor recurrence within 6 months of receiving adjuvant therapy. The US Food and Drug Administration (FDA) approval was based on the results of the EMILIA study, a phase 3 clinical trial that compared treatment with trastuzumab emtansine vs capecitabine + lapatinib in participants with HER2+, locally advanced, or metastatic BC. This trial emerged from the need for well-tolerated, HER2-directed therapies for patients with this type of cancer. Trastuzumab emtansine consists of trastuzumab, a mAb that targets HER2 (which is overexpressed in about 20% of BCs), linked to emtansine, a cytotoxic payload that inhibits cell division. The trastuzumab emtansine group had a median overall survival (OS) of 29.9 months vs 25.9 months in the capecitabine + lapatinib group, for a hazard ratio (HR) of 0.75 (95% CI: 0.64, 0.88).
Another ADC, sacituzumab govitecan, targets the Trop-2 protein, which is overexpressed in BC. This ADC includes a mAb that is linked to SN-38, a cytotoxic payload that inhibits DNA replication. Triple-negative breast cancer (TNBC) is a subtype of BC that does not have receptors for estrogen, progesterone, or HER2—making it more difficult to treat than other forms of BC. Sacituzumab govitecan is used to treat patients with metastatic TNBC who have received at least 2 prior therapies for metastatic disease. Sacituzumab govitecan is also approved for the treatment of patients with unresectable locally advanced or metastatic hormone-receptor–positive (HR+), and HER2-negative (HER2−) BC who have received endocrine-based therapy and at least 2 additional systemic therapies in the metastatic setting. Sacituzumab govitecan was the first Trop-2–directed ADC to demonstrate OS benefit in patients with HR+/HER2− mBC who had received prior endocrine-based therapy and at least 2 chemotherapies. It is now also recommended as a Category 1 preferred treatment for metastatic HR+/HER2− BC by the National Comprehensive Cancer Network.
The results of the TROPiCS-02 study, which led to the FDA approval of sacituzumab govitecan, demonstrated a median OS of 14.4 months with sacituzumab govitecan vs 11.2 months with treatment of physician’s choice (HR, 0.79; 95% CI: 0.65, 0.96; P = 0.02). This represents a 3.2-month improvement in survival and a 21% reduction in the risk for patient death. Before this medicine was approved, there were limited options to offer patients with BC after endocrine-based therapy and chemotherapy.
A third ADC, trastuzumab deruxtecan, targets the HER2 protein, like trastuzumab emtansine, but with a different cytotoxic payload. It consists of trastuzumab linked to deruxtecan, whose cytotoxicity inhibits DNA replication. It is approved for the treatment of HER2+ mBC. Its FDA approval was based on the results of the DESTINY-Breast04 phase 3 clinical trial, which demonstrated that treatment with trastuzumab deruxtecan, when compared with standard-of-care chemotherapy, doubles progression-free survival among patients with mBC that expresses low levels of HER2. The median OS for patients in the HR+ group who received trastuzumab deruxtecan was 23.9 months vs 17.5 months for those who received chemotherapy. In the hormone receptor-negative (HR−) group, the median OS for those who took trastuzumab deruxtecan was 16.6 months vs 10.3 months for those treated with chemotherapy.
The emergence of ADCs have demonstrated promising advancements in the treatment of BC, particularly in patients with HER2+ or triple-negative disease. ADCs have given new hope to and prolonged life for patients living with pretreated HR+/HER2− mBC. ADCs also have the potential to provide a more effective and less toxic treatment option for patients with BC. However, further research is needed to fully understand their long-term effects and to develop new ADCs that target other types of BC.
Surgery for early breast cancer can worsen frailty in older women
according to a new study.
About 1 in 5 experienced clinically significant deterioration in frailty status after treatment, the study team found. Women at highest risk for declines in frailty following treatment had “robust” baseline frailty status at diagnosis and underwent more invasive mastectomy compared with lumpectomy.
The fact that “robust” older women were more likely to become frail after locoregional therapy suggests that “thoughtful treatment decisions should be undertaken in all older women, not simply those who have frailty at diagnosis,” said the investigators, led by Christina Minami, MD, of Dana-Farber/Brigham and Women’s Cancer Center in Boston.
The study findings emphasize that there is no one-size-fits-all approach to breast cancer treatment in the elderly, said Sarah P. Cate, MD, director, Breast Surgery Quality Program, Mount Sinai Health System, New York, who wasn’t involved in the research. “Some patients will sail through a surgery, and others are severely affected by it.”
The study was published online in JAMA Surgery.
Given the growing number of older adults with breast cancer, understanding how age-related syndromes, such as frailty, may alter cancer outcomes and how cancer treatments change aging trajectories remains important.
To investigate, Dr. Minami and colleagues used Surveillance, Epidemiology, and End Results Medicare data to identify 31,084 women (mean age, 73) who had been diagnosed with ductal carcinoma in situ (DCIS) or stage I HR-positive, ERBB2-positive breast cancer and who underwent surgery (23% mastectomy, 77% lumpectomy) and radiation therapy.
Worsening frailty status was defined as a decline of 0.03 or greater in a validated frailty index from the time of diagnosis to 1 year. This level of change has been linked to greater mortality risk and greater cost of care.
Frailty status at diagnosis was “robust” in 56% of the women, prefrail in 40%, mildly frail in 4%, and moderately to severely frail in 0.3%.
According to the researchers, 21.4% of the women experienced clinically significant declines in their frailty status after treatment. These declines occurred in 25% of women who underwent mastectomy and 20% of those who underwent lumpectomy.
After adjusting for covariates, there was a higher likelihood of worsening frailty among women who were robustly frail at baseline, in comparison with those who were moderately to severely frail at baseline (odds ratio, 6.12), and in those who underwent mastectomy vs. lumpectomy (OR, 1.31).
Older age and race were also linked to worsening frailty status following treatment. Compared with younger women (aged 65-74 years), older women were more likely to experience worsening frailty (OR, 1.21 for women aged 75-79; OR, 1.53 for those aged 80-84; OR, 1.94 for those aged 85 and older). In addition, Black women were more likely than non-Hispanic White women to experience worsening frailty after treatment (OR, 1.12).
“Previous studies have documented lasting declines in functional status after surgery in older patients with breast cancer, but breast cancer treatment has not been implicated in worsening frailty to date,” Dr. Minami and colleagues explain. But “given the substantial proportion of women experiencing worsening frailty and the significant difference by breast surgery type, frailty status as a cancer therapy outcome should be further explored.” In addition, “tailoring locoregional therapy intensity in this population is important,” they write.
Dr. Cate explained that randomized clinical trials such as COMET and LORIS, which explore the monitoring of patients with DCIS in lieu of active treatment, “will likely make a big impact on this population, as we currently do not have randomized controlled data for observation of breast cancer.”
Dr. Cate added as well that assessing a patient’s ECOG [Eastern Cooperative Oncology Group] performance status is vital “to determine who can really tolerate a breast cancer surgery” and that opting for antiestrogens, such as aromatase inhibitors, which can keep cancer at bay for years, “may be preferable for many older patients.”
The study was funded by Brigham and Women’s Hospital’s Department of Surgery’s Beal Fellowship. Dr. Minami and Dr. Cate have disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
according to a new study.
About 1 in 5 experienced clinically significant deterioration in frailty status after treatment, the study team found. Women at highest risk for declines in frailty following treatment had “robust” baseline frailty status at diagnosis and underwent more invasive mastectomy compared with lumpectomy.
The fact that “robust” older women were more likely to become frail after locoregional therapy suggests that “thoughtful treatment decisions should be undertaken in all older women, not simply those who have frailty at diagnosis,” said the investigators, led by Christina Minami, MD, of Dana-Farber/Brigham and Women’s Cancer Center in Boston.
The study findings emphasize that there is no one-size-fits-all approach to breast cancer treatment in the elderly, said Sarah P. Cate, MD, director, Breast Surgery Quality Program, Mount Sinai Health System, New York, who wasn’t involved in the research. “Some patients will sail through a surgery, and others are severely affected by it.”
The study was published online in JAMA Surgery.
Given the growing number of older adults with breast cancer, understanding how age-related syndromes, such as frailty, may alter cancer outcomes and how cancer treatments change aging trajectories remains important.
To investigate, Dr. Minami and colleagues used Surveillance, Epidemiology, and End Results Medicare data to identify 31,084 women (mean age, 73) who had been diagnosed with ductal carcinoma in situ (DCIS) or stage I HR-positive, ERBB2-positive breast cancer and who underwent surgery (23% mastectomy, 77% lumpectomy) and radiation therapy.
Worsening frailty status was defined as a decline of 0.03 or greater in a validated frailty index from the time of diagnosis to 1 year. This level of change has been linked to greater mortality risk and greater cost of care.
Frailty status at diagnosis was “robust” in 56% of the women, prefrail in 40%, mildly frail in 4%, and moderately to severely frail in 0.3%.
According to the researchers, 21.4% of the women experienced clinically significant declines in their frailty status after treatment. These declines occurred in 25% of women who underwent mastectomy and 20% of those who underwent lumpectomy.
After adjusting for covariates, there was a higher likelihood of worsening frailty among women who were robustly frail at baseline, in comparison with those who were moderately to severely frail at baseline (odds ratio, 6.12), and in those who underwent mastectomy vs. lumpectomy (OR, 1.31).
Older age and race were also linked to worsening frailty status following treatment. Compared with younger women (aged 65-74 years), older women were more likely to experience worsening frailty (OR, 1.21 for women aged 75-79; OR, 1.53 for those aged 80-84; OR, 1.94 for those aged 85 and older). In addition, Black women were more likely than non-Hispanic White women to experience worsening frailty after treatment (OR, 1.12).
“Previous studies have documented lasting declines in functional status after surgery in older patients with breast cancer, but breast cancer treatment has not been implicated in worsening frailty to date,” Dr. Minami and colleagues explain. But “given the substantial proportion of women experiencing worsening frailty and the significant difference by breast surgery type, frailty status as a cancer therapy outcome should be further explored.” In addition, “tailoring locoregional therapy intensity in this population is important,” they write.
Dr. Cate explained that randomized clinical trials such as COMET and LORIS, which explore the monitoring of patients with DCIS in lieu of active treatment, “will likely make a big impact on this population, as we currently do not have randomized controlled data for observation of breast cancer.”
Dr. Cate added as well that assessing a patient’s ECOG [Eastern Cooperative Oncology Group] performance status is vital “to determine who can really tolerate a breast cancer surgery” and that opting for antiestrogens, such as aromatase inhibitors, which can keep cancer at bay for years, “may be preferable for many older patients.”
The study was funded by Brigham and Women’s Hospital’s Department of Surgery’s Beal Fellowship. Dr. Minami and Dr. Cate have disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
according to a new study.
About 1 in 5 experienced clinically significant deterioration in frailty status after treatment, the study team found. Women at highest risk for declines in frailty following treatment had “robust” baseline frailty status at diagnosis and underwent more invasive mastectomy compared with lumpectomy.
The fact that “robust” older women were more likely to become frail after locoregional therapy suggests that “thoughtful treatment decisions should be undertaken in all older women, not simply those who have frailty at diagnosis,” said the investigators, led by Christina Minami, MD, of Dana-Farber/Brigham and Women’s Cancer Center in Boston.
The study findings emphasize that there is no one-size-fits-all approach to breast cancer treatment in the elderly, said Sarah P. Cate, MD, director, Breast Surgery Quality Program, Mount Sinai Health System, New York, who wasn’t involved in the research. “Some patients will sail through a surgery, and others are severely affected by it.”
The study was published online in JAMA Surgery.
Given the growing number of older adults with breast cancer, understanding how age-related syndromes, such as frailty, may alter cancer outcomes and how cancer treatments change aging trajectories remains important.
To investigate, Dr. Minami and colleagues used Surveillance, Epidemiology, and End Results Medicare data to identify 31,084 women (mean age, 73) who had been diagnosed with ductal carcinoma in situ (DCIS) or stage I HR-positive, ERBB2-positive breast cancer and who underwent surgery (23% mastectomy, 77% lumpectomy) and radiation therapy.
Worsening frailty status was defined as a decline of 0.03 or greater in a validated frailty index from the time of diagnosis to 1 year. This level of change has been linked to greater mortality risk and greater cost of care.
Frailty status at diagnosis was “robust” in 56% of the women, prefrail in 40%, mildly frail in 4%, and moderately to severely frail in 0.3%.
According to the researchers, 21.4% of the women experienced clinically significant declines in their frailty status after treatment. These declines occurred in 25% of women who underwent mastectomy and 20% of those who underwent lumpectomy.
After adjusting for covariates, there was a higher likelihood of worsening frailty among women who were robustly frail at baseline, in comparison with those who were moderately to severely frail at baseline (odds ratio, 6.12), and in those who underwent mastectomy vs. lumpectomy (OR, 1.31).
Older age and race were also linked to worsening frailty status following treatment. Compared with younger women (aged 65-74 years), older women were more likely to experience worsening frailty (OR, 1.21 for women aged 75-79; OR, 1.53 for those aged 80-84; OR, 1.94 for those aged 85 and older). In addition, Black women were more likely than non-Hispanic White women to experience worsening frailty after treatment (OR, 1.12).
“Previous studies have documented lasting declines in functional status after surgery in older patients with breast cancer, but breast cancer treatment has not been implicated in worsening frailty to date,” Dr. Minami and colleagues explain. But “given the substantial proportion of women experiencing worsening frailty and the significant difference by breast surgery type, frailty status as a cancer therapy outcome should be further explored.” In addition, “tailoring locoregional therapy intensity in this population is important,” they write.
Dr. Cate explained that randomized clinical trials such as COMET and LORIS, which explore the monitoring of patients with DCIS in lieu of active treatment, “will likely make a big impact on this population, as we currently do not have randomized controlled data for observation of breast cancer.”
Dr. Cate added as well that assessing a patient’s ECOG [Eastern Cooperative Oncology Group] performance status is vital “to determine who can really tolerate a breast cancer surgery” and that opting for antiestrogens, such as aromatase inhibitors, which can keep cancer at bay for years, “may be preferable for many older patients.”
The study was funded by Brigham and Women’s Hospital’s Department of Surgery’s Beal Fellowship. Dr. Minami and Dr. Cate have disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM JAMA SURGERY